CN109731105B - Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof - Google Patents

Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof Download PDF

Info

Publication number
CN109731105B
CN109731105B CN201910019951.6A CN201910019951A CN109731105B CN 109731105 B CN109731105 B CN 109731105B CN 201910019951 A CN201910019951 A CN 201910019951A CN 109731105 B CN109731105 B CN 109731105B
Authority
CN
China
Prior art keywords
nano
autophagy
nasal cavity
cis
neurodegenerative diseases
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201910019951.6A
Other languages
Chinese (zh)
Other versions
CN109731105A (en
Inventor
刘刚
刘静怡
文磊
张金凤
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nafi (Shenzhen) Pharmaceutical Technology Co., Ltd
Original Assignee
Nafi Shenzhen Pharmaceutical Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nafi Shenzhen Pharmaceutical Technology Co ltd filed Critical Nafi Shenzhen Pharmaceutical Technology Co ltd
Priority to CN201910019951.6A priority Critical patent/CN109731105B/en
Priority to CN202110665044.6A priority patent/CN113384541B/en
Publication of CN109731105A publication Critical patent/CN109731105A/en
Priority to PCT/CN2020/070987 priority patent/WO2020143688A1/en
Application granted granted Critical
Publication of CN109731105B publication Critical patent/CN109731105B/en
Priority to US17/372,403 priority patent/US20220000804A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/12Aerosols; Foams
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Psychology (AREA)
  • Otolaryngology (AREA)
  • Dispersion Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pulmonology (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and a preparation method thereof, and the nasal cavity nano autophagy inducer comprises hydrophobic micromolecules with autophagy induction effect and an amphiphilic surfactant; firstly, preparing a good solvent solution, then preparing a self-contained carrier-free nano particle suspension emulsion by a reprecipitation method, and freeze-drying to prepare a freeze-dried powder; before use, the freeze-dried powder is re-suspended in isotonic physiological saline to obtain the traditional Chinese medicine. According to the nasal cavity nano autophagy inducer, under the nasal cavity administration route, a medicine can be absorbed by olfactory mucosa, firstly reaches and is enriched in olfactory related regions of a brain, and the nasal cavity nano autophagy inducer has a special effect of relieving common symptoms of olfactory disturbance of early neurodegenerative diseases; has obvious clearing effect on abnormal protein aggregation in olfactory region and other pathological change regions, and has important significance for preventing further deterioration of early AD, PD and other neurodegenerative diseases. The inducer has no carrier, no biodegradation problem and accumulative toxicity, and the drug loading rate is up to more than 25%.

Description

Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof
Technical Field
The invention belongs to the field of biological medicines, and relates to a nasal cavity nano autophagy inducer and a preparation method thereof. In particular to a nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and a preparation method thereof.
Background
Autophagy is a process of phagocytosing self cytoplasmic proteins or organelles, coating the proteins or organelles into vesicles, fusing with lysosomes and degrading the contents coated by the proteins or organelles, so that the metabolic needs of cells and the renewal of the organelles are realized. Neurons are heavily dependent on autophagy. Disruption of the autophagy pathway can lead to accumulation of ubiquitinated protein aggregates within neurons, inducing neuronal degeneration, and subsequently leading to the development of neurodegenerative diseases.
Alzheimer's Disease (AD) is the most common neurodegenerative disease, two of the main features of which, senile plaque formation due to amyloid β peptide accumulation and neurofibrillary tangle formation due to Tau aggregation, can be effectively degraded by activation of autophagy pathways. The prevalence rate of the Parkinson's Disease (PD) in people over 60 years old is 1%, and the PD is mainly manifested by motor dysfunction such as tremor and the like, which is caused by the degeneration of dopaminergic neurons, the pathogenesis of the PD is directly related to the defect of an autophagosomal pathway, and the main pathological marker is an abnormal aggregate Lewy body of alpha-synuclein (a-synuclin).
According to a plurality of clinical reports, the olfactory disorder is a common symptom in the early stage of neurodegenerative diseases such as AD and PD, more than ninety percent of Parkinson patients are accompanied by the olfactory disorder, and the olfactory disorder can be used as an index for identifying PD and atypical Parkinson syndrome, and provides help for early diagnosis and differential diagnosis of PD. The mechanism of olfactory impairment of neurodegenerative diseases is associated with the accumulation of toxic proteins in the olfactory region. In pathological conditions, autophagy pathways in olfactory-related regions are damaged, resulting in abnormal degradation of proteins, accumulation of pressure on neurons, and induction of disease progression. For the abnormal protein aggregation, no specific medicine exists clinically at present. For example, the treatment of PD can only alleviate symptoms by supplementing dopamine, and does not have a therapeutic effect on the disease. And as the disease worsens, dopamine therapy becomes progressively ineffective, with serious side effects. Therefore, the development of new therapeutic drugs is urgently required.
The autophagy inducer can enhance the occurrence of autophagy flow in neurons through various mechanisms, such as increasing the generation of autophagosome, promoting the fusion of autophagosome and lysosome, enhancing the function of lysosome, increasing the quantity of lysosome and the like, repairing damaged autophagy channels, promoting the degradation of toxic proteins, and reducing nerve damage caused by accumulation of toxic proteins. Certain autophagy inducers can activate the autophagy pathway by binding to TFEB proteins and are potent autophagy inducers. However, the organic micromolecules with strong hydrophobicity have poor drug forming property, and the oral administration has low bioavailability, less brain enrichment and short in vivo circulation time, so that various problems limit the exploration research on pathological models such as AD and PD and the application of the organic micromolecules in future clinical transformation.
With the rapid development of nanotechnology, the nanometer preparation has been widely used in the medical and biological fields because of its advantages of protecting the drug from being destroyed, prolonging the effective drug retention time, controlling the drug release, and reducing the toxic and side effects of the drug. Many reports show that the drug-loading rate of the drug nanoparticles encapsulated by carriers such as liposome and polymer is usually lower than 10%, and the enrichment of the carriers such as polymer in the brain may bring about potential toxic and side effects, which is a troublesome problem that further application of the nanoparticles to clinic is hindered. In addition, the blood brain barrier is an important physiological barrier, which prevents more than 98% of drugs from entering brain tissues, and particularly, nano drugs with the diameter of about 100nm are more difficult to penetrate the blood brain barrier to reach the nervous system to play a role. Therefore, the development of a high drug-loading, high brain targeting, safe and nontoxic nano delivery system for the treatment of nervous system diseases of autophagy small molecule drugs is urgently needed.
Disclosure of Invention
The invention mainly aims to provide a nasal cavity nano autophagy inducer and a preparation method thereof, in particular to a self-carried carrier-free nasal cavity nano autophagy inducer and a preparation method thereof, which are applied to early neurodegenerative diseases.
The technical scheme of the invention is as follows:
a nasal cavity nanometer autophagy inducer for preventing and treating early neurodegenerative diseases comprises hydrophobic small molecules with autophagy induction effect and amphiphilic surfactant; firstly, preparing a good solvent solution of 1-10mg/mL and 0.5-5mg/mL of an autophagy inducing drug of an amphiphilic surfactant, and then dropwise adding the good solvent solution into deionized water, wherein the volume ratio of the good solvent solution to the deionized water is (0.5-5): 50, air blowing is assisted while dropwise adding, and good solvent volatilization is assisted; preparing self-carried carrier-free nano particle suspension emulsion with the particle size of 50-200nm by a reprecipitation method, and freeze-drying to prepare freeze-dried powder; before use, the freeze-dried powder is re-suspended in isotonic physiological saline to obtain the self-carried non-carrier nasal cavity nano autophagy inducer.
The autophagy inducer of the invention can clear abnormal protein aggregation by inducing autophagy.
Preferably, the surface potential of the self-carrying unsupported nanoparticles is from-10 to-60 mV. More preferably-10 to-30 mV.
Preferably, the early neurodegenerative disease includes alzheimer's disease and parkinson's disease.
The early neurodegenerative disease is also accompanied by olfactory disorder symptoms, and the nasal cavity nano autophagy inducer is an autophagy inducer with high targeting enrichment at olfactory bulb parts. The nasal cavity nano autophagy inducer has obvious clearing effect on abnormal protein aggregation in olfactory region and other lesion regions.
Preferably, the hydrophobic small molecule is curcumin analogue of the following structural formula, cis isomer thereof or mixture of the two in any proportion:
Figure GDA0003017689500000021
preferably, the hydrophobic small molecule is a mixture of the curcumin analogue and cis-isomer thereof, and the weight ratio of the cis-isomer in the mixture accounts for 25-35% of the total mixture.
Preferably, the mixture of cis-isomers with a weight ratio of 25-35% of the total mixture is prepared by subjecting a methanol solution of curcumin analogues to ultraviolet irradiation for 1.5-2.5 h.
Preferably, the methanol solution concentration of the curcumin analogue is 0.5 to 5mg/ml, and more preferably 0.5 to 1.5 mg/ml. If the time of the ultraviolet irradiation is shorter than 1.5 hours, the cis-isomer cannot be formed in a sufficient amount, and if it is longer than 2.5 hours, the by-production of the by-product starts, and the ultraviolet irradiation is particularly preferably 2 hours.
Preferably, the nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases further comprises oligosaccharide, wherein the concentration of the chitosan oligosaccharide in isotonic physiological saline solution is 0.01-0.2% (w/v).
In the present invention, the amphiphilic surfactant is not limited, and may be any pharmaceutically acceptable surfactant having a lipophilic group and a hydrophilic group and capable of forming a self-assembled nanoparticle structure with the autophagy-inducing drug molecule of the present invention. Preferably, the amphiphilic surfactant is a polyethylene glycol derivative, and more preferably, carboxyl polyethylene glycol or polymaleic anhydride 18 carbene-polyethylene glycol.
Preferably, the gas is nitrogen or an inert gas, preferably nitrogen. And blowing gas to assist the good solvent to volatilize so as to ensure the formation of nano particles and prevent potential safety hazards caused by solvent residue.
Preferably, the nasal cavity nano autophagy inducer has an average particle size of 50-200nm, preferably 50-150nm, more preferably 50-120 nm.
Preferably, the drug loading rate of the nasal cavity nano autophagy inducer is more than 25%.
Preferably, the nasal cavity nano autophagy inducer further comprises chitosan oligosaccharide, the concentration of the chitosan oligosaccharide in isotonic physiological saline solution is 0.01-0.2% (w/v), and when the nasal cavity nano autophagy inducer is used, the freeze-dried powder is re-suspended in the isotonic physiological saline containing the chitosan oligosaccharide.
Chitosan oligosaccharide, also called chitosan oligosaccharide and oligochitosan, has molecular weight less than 3200Da, has the unique functions of high solubility, complete water solubility, easy absorption and utilization by organisms and the like which chitosan does not have. The chitosan oligosaccharide is nontoxic functional low molecular weight amino sugar and has a polycation structure, and can be modified outside nanoparticles to prevent irritation of drugs to the environment in nasal cavity; the chitosan oligosaccharide can easily act with negatively charged groups on the surface of mucosal cells, can change the fluidity and permeability of cell membranes and increase the absorption of nanoparticles, and has certain immunoregulation and neuroprotection effects, and the effect of the chitosan oligosaccharide is 14 times that of chitosan.
The polymerization degree of the chitosan oligosaccharide used in the invention is 2-20, or the molecular weight is less than or equal to 3200 Da.
Preferably, the oligosaccharide of the present invention has a concentration of 0.01-0.2% (w/v) in isotonic physiological saline solution, and if less than 0.01%, it is difficult to play a role in increasing absorption, preventing irritation, etc., and if more than 0.2%, it easily causes aggregation of negatively charged nanoparticles.
Re-suspending the freeze-dried powder in isotonic physiological saline, wherein the concentration of the freeze-dried powder can be prepared according to the requirement. Preferably 3-7 mg/ml.
The formation of the nano particles is influenced by the molecular structure of hydrophobic small molecules, and non-covalent binding force exists among the molecules, so that the difference of the nano structures can be caused due to the difference of the molecular configurations. In order to enhance the stability of the nano-particles, the invention adds the amphiphilic surfactant before solvent exchange, uniformly mixes the amphiphilic surfactant and hydrophobic micromolecules in an organic solvent according to a certain proportion, then carries out solvent exchange, and obtains the nano-particles without carrier coating by a reprecipitation method.
The nasal cavity nano autophagy inducer of the invention has no other carrier components, so the nasal cavity nano autophagy inducer has high drug loading, low toxicity, good safety, small and uniform particle size, high stability and long circulation time in vivo.
In the invention, the medicament contained in the nasal cavity nano autophagy inducer is a hydrophobic organic micromolecule with autophagy induction effect, and can be used for preventing and treating neurodegenerative diseases after the medicament forming property is improved.
The invention can also be added with an antioxidant which can be one or more of sodium metabisulfite, sodium bisulfite, sodium sulfite, sodium thiosulfate, cysteine hydrochloride, vitamin C, vitamin E and thiourea, and the dosage of the antioxidant is the conventional dosage specified in pharmaceutics.
The invention can also be added with preservative which can be one or more of methyl p-hydroxybenzoate, ethyl p-hydroxybenzoate, propyl p-hydroxybenzoate, butyl p-hydroxybenzoate, benzalkonium bromide, benzalkonium chloride, chlorobutanol, phenethyl alcohol, thimerosal, phenylmercuric nitrate, sorbic acid and chlorhexidine, and the dosage of the preservative is conventional dosage specified in pharmaceutics.
The invention can also be added with an osmotic pressure regulator, wherein the osmotic pressure regulator can be one or more of sodium chloride, glucose, lactose and mannitol, and the dosage of the osmotic pressure regulator is the conventional dosage specified in pharmaceutics.
The invention provides a self-carried carrier-free nasal cavity nano autophagy inducer, which is characterized in that nano particles are spherical or approximately spherical, the surface potential is negative electricity, hydrophobic autophagy inducing drug molecules are prepared into nano particle suspension emulsion through a reprecipitation method, and freeze drying is carried out to obtain nano particle freeze-dried powder. Before use, the freeze-dried powder is re-suspended in isotonic physiological saline to obtain the nasal cavity nano autophagy inducer, and the nasal cavity nano autophagy inducer is administrated by nasal drip or nasal spray and is used for treating neurodegenerative diseases.
The delivery medicament is a hydrophobic small molecule with autophagy induction effect, the delivery mode is a self-carried carrier-free nasal cavity nano delivery system, a nasal drop or spray mode is adopted, the nasal cavity nano delivery system is non-invasive, carrier-free, biodegradation problems and accumulated toxicity are avoided, the medicament carrying amount is over 25 percent, the binding capacity of the small molecule and a target receptor is highly reserved, the small molecule medicament can be slowly released in a neuron in a pH response manner, and an autophagy channel is specifically activated, so that the intracerebral toxic protein accumulation of the neurodegenerative disease is efficiently eliminated, particularly, the nasal cavity nano autophagy inducer has a special effect on early olfactory disturbance of the disease, and has important significance for preventing further deterioration of the disease.
In a second aspect, the present invention provides a method for preparing a nasal nano autophagy inducer according to the first aspect, the method comprising the steps of:
1) firstly, preparing a good solvent solution of 1-10mg/mL and 0.5-5mg/mL of an amphiphilic surfactant of a hydrophobic micromolecular drug, and then dropwise adding the good solvent solution into deionized water: the volume ratio of the good solvent solution to the deionized water is (0.5-5): 50, dripping while blowing gas and volatilizing good solvent, 2) preparing self-carried carrier-free nano particle suspension emulsion with the particle size of 50-200nm by a reprecipitation method, and freeze-drying to prepare freeze-dried powder;
3) before use, the freeze-dried powder is re-suspended in isotonic physiological saline to obtain the nasal cavity nano autophagy inducer.
The method for preparing the nasal cavity nano autophagy inducer is carried out by a reprecipitation method, when a good solvent is converted into water (a poor solvent), hydrophobic small molecules with autophagy induction function are separated out to form nano particles, and an amphiphilic surfactant is added, so that the stability and water dispersibility of the nasal cavity nano autophagy inducer can be further enhanced. The method is simple and easy to implement, does not need complex operation and conditions, and can be carried out at room temperature.
It should be emphasized that, in the present invention, the amphiphilic surfactant is added before the nanoparticles are formed, i.e. dissolved in a good solvent together with the small molecules, and after the mixture is mixed uniformly, the aqueous phase is added dropwise to prepare the nanoparticles. During the dropwise addition, a gas (preferably nitrogen) is blown to remove the organic solvent. The method is different from the method of firstly forming nano particles and then adding the amphiphilic surfactant for surface modification, and the product is also different.
In the method for preparing the nasal cavity nano autophagy inducer, the good solvent is mutually soluble with water. According to the Flory-Krigboum dilute solution theory, a good solvent refers to a solvent with a solute interaction parameter of less than 0.5.
Preferably, the good solvent is one or more of acetone, methanol, ethanol and tetrahydrofuran, and more preferably tetrahydrofuran.
In the method for preparing the nasal cavity nano autophagy inducer, the water can be deionized water, distilled water or double distilled water, and the like, and the deionized water is preferred.
In the method for preparing the nasal cavity nano autophagy inducer, the concentration of the hydrophobic drug molecules dissolved in the good solvent in the step (1) is 0.5-5mg/mL, and preferably 1 mg/mL.
In the method for preparing the nasal cavity nano autophagy inducer, the volume ratio of the good solvent to the water in the step (1) is preferably (1-3): 50, e.g. 1: 50. 1.2: 50. 1.5: 50. 1.8: 50. 1.9: 50. 2.1: 50. 2.5: 50 or 2.8: 50, preferably 2: 50.
preferably, the reaction temperature in step (1) is 20 to 30 ℃, more preferably 25 ℃.
In the method for preparing a nasal cavity nano autophagy inducer of the invention, the concentration of the amphiphilic surfactant added in the step (2) in the good solvent may be 1-10mg/mL, such as 1mg/mL, 2mg/mL, 3mg/mL, 4mg/mL, 5mg/mL, 6mg/mL, 7mg/mL, 8mg/mL, 9mg/mL or 10mg/mL, preferably 2 mg/mL.
Preferably, the time of ultrasonic dispersion in step (2) is 3-30min, such as 3min, 4min, 5min, 8min, 10min, 15min, 20min, 25min or 28min, more preferably 5 min.
The self-assembled nano particles can be aggregated when being stored in an aqueous solution for a long time, the stability of the self-assembled nano particles can be obviously improved by freeze-drying the self-assembled nano particles, and the freezing temperature is 10-20 ℃ lower than the eutectic point of the nano particles and water, and the freeze-drying is carried out for 24-90h, preferably 48h, under the pressure of 10 Pa.
In order to avoid aggregation and particle size change of the freeze-dried nanoparticles, a cryoprotectant, such as glucose, mannitol, lactose, NaCl and the like, is added firstly, so that a large amount of micro ice crystals are promoted to form during freezing, or a freeze-dried product is in a loose state, and the nanoparticles can be favorably kept in the original shape and can be easily re-dispersed in water.
The nasal cavity nano autophagy inducer can be administrated by a nasal route in a spray or nasal drop mode.
The invention has the beneficial effects that:
(1) according to the nasal cavity nano autophagy inducer, under the nasal cavity administration route, a medicine can be absorbed by olfactory mucosa, firstly reaches and is enriched in olfactory related regions of a brain, and the nasal cavity nano autophagy inducer has a special effect of relieving common symptom olfactory disturbance of early neurodegenerative diseases; has obvious clearing effect on abnormal protein aggregation in olfactory region and other pathological change regions, and has important significance for preventing further deterioration of early AD, PD and other neurodegenerative diseases.
(2) The invention relates to a nasal cavity nano autophagy inducer delivery system which is high in drug loading capacity, high in brain targeting property, safe and non-toxic, is applied to hydrophobic organic small molecules with autophagy induction effects, and is simple to operate, wide in application range and strong in universality. Compared with the small-molecule prodrug, the prodrug has the advantages of remarkably improving water dispersibility and drug forming property, enhancing bioavailability, reducing administration frequency, reducing toxic and side effects and the like. Compared with the traditional liposome or polymer nano drug-carrying system, the nano system has no carrier, no biodegradation problem and accumulative toxicity, the drug-carrying rate is up to more than 25 percent, the binding capacity of the target receptor of the original molecule is highly retained, the nano system has the drug release characteristic of pH responsiveness, can exert the effect in a long-acting and slow-release manner, and has good application prospect in the treatment of nervous system diseases.
(2) The nasal cavity nano autophagy inducer provided by the invention has the advantages that the medicine is efficiently delivered into the brain by bypassing the blood brain barrier along the olfactory nerve and other ways through the direct nasal brain passage, the gastrointestinal tract degradation and liver first-pass effect can be avoided, the brain targeting property is obviously enhanced, the nasal cavity nano autophagy inducer has the characteristics of high bioavailability, quick response, good patient compliance and the like, the visceral enrichment of a peripheral circulatory system can be reduced, and the potential side effect of long-term administration is reduced. Compared with oral medicine, the nasal preparation has no first-pass effect and reduced medicine loss. Compared with intravenous injection, nasal administration only needs nasal drip, spray and other modes, is convenient to use and has no wound, and the compliance of patients is improved. Especially for the patients with neurodegenerative diseases who take medicines for a long time, the pain of the patients can be relieved, the compliance of the patients is good, the patients can take medicines by themselves conveniently, the risk brought by long-term medicine taking is reduced, and the application prospect is good.
Drawings
The invention is further illustrated by the following figures and examples.
FIG. 1 is a Scanning Electron Microscope (SEM) image of a nasal nano autophagy inducer. Wherein a is the M1 nanoparticles prepared in example 1; b is the M1 nanoparticles loaded with the TPAAQ probe prepared in example 2.
FIG. 2 is a Transmission Electron Microscope (TEM) image of a nasal nano autophagy inducer. Wherein a is the M1 nanoparticles prepared in example 1; b is the M1 nanoparticles loaded with the TPAAQ probe prepared in example 2.
FIG. 3 shows the particle size distribution of the nasal nano autophagy inducer. Wherein a is the M1 nanoparticles prepared in example 1; b is the M1 nanoparticles loaded with the TPAAQ probe prepared in example 2.
Figure 4 is a graph of drug loading rate of nasal nano autophagy inducer. Wherein a is the M1 nanoparticles prepared in example 1; b is the M1 nanoparticles loaded with the TPAAQ probe prepared in example 2.
FIG. 5 is a surface potential diagram of M1 nanoparticles prepared in example 1.
FIG. 6 is a Tyndall effect optical characteristic diagram of M1 nanoparticles prepared in example 1.
Fig. 7 pH-responsive drug release profile of M1 nanoparticles prepared in example 1.
Fig. 8 cytotoxicity of M1 nanoparticles prepared in example 1 with small molecule M1 drug.
Fig. 9 is a graph of the in vitro neuroprotective effect of M1 nanoparticles prepared in example 1.
FIG. 10 is a graph showing the binding effect of M1 nanoparticles prepared in example 1 to TFEB protein, a target protein.
FIG. 11. M1 nanoparticles prepared in example 1 induced autophagy flow fluorescence profiles.
FIG. 12 cell uptake plots of M1 nanoparticles loaded with TPAAQ fluorescent probes prepared in example 2.
FIG. 13 is a graph of M1 drug content in mouse brain and plasma after nasal brain administration of M1 nanoparticles of example 3.
FIG. 14 shows fluorescence biodistribution of brain and organs of mice after transnasal administration of M1 nanoparticles of TPAAQ fluorescent probe of example 4.
FIG. 15 is the open field behavioral graph of the Parkinson model mouse of example 5.
FIG. 16 is a gait behavior diagram of the Parkinson model mouse of example 5.
Fig. 17 olfactory bulb and striatum, substantia nigra profiles of the parkinsonian model mouse of example 5 after nasal administration of M1 nanoparticles, wherein the M1 nanoparticles are indicated by arrows.
FIG. 18 is a graph showing the test of the expression levels of toxic proteins and proteins associated with the autophagy pathway in olfactory region of mice belonging to the Parkinson's model of example 5.
FIG. 19 is a graph showing the test of the expression level of toxic proteins and autophagy pathway-associated proteins in the substantia nigra of mice in the Parkinson's model of example 5.
FIG. 20 spectrum of 294.34 molecular weight compound in mass spectrum of example 1
Figure 21 shows curcumin analog conversion in different conditions of example 1.
Detailed Description
Embodiments of the present invention will be described in detail with reference to examples. It will be appreciated by those skilled in the art that the following examples are only preferred embodiments of the invention to facilitate a better understanding of the invention and should not be taken as limiting the scope of the invention. Various modifications and changes may be made by those skilled in the art, and any modification, equivalent replacement or improvement made without departing from the spirit and principle of the present invention should be covered within the protection scope of the present invention. The experimental methods in the following examples are all conventional methods unless otherwise specified; the experimental materials used, unless otherwise specified, were purchased from conventional biochemical manufacturers.
EXAMPLE 1 preparation of the Mixed isomer M1
The mixed isomer is an isomer mixture of curcumin analogues of the following structural formula:
Figure GDA0003017689500000081
according to the results of the computer simulations of the applicant, the higher the proportion of cis-isomers in the mixture, the greater the biological activity of the product. In practice, however, the higher the product, the higher the yield of by-products.
In this example, a process was provided in which 30% conversion of the isomeric product was obtained, the process was simple, and no by-product was produced.
According to the hydrophobic property of the curcumin analogue, the curcumin analogue is dissolved in a good solvent, and different degrees of isomer conversion can occur under different irradiation conditions, so that cis-trans isomer mixtures of the curcumin analogue with different proportions are obtained. Wherein the conversion rate of sunlight irradiation is the highest, but byproducts are generated; the conversion rate is ultraviolet irradiation and radioactive iodine irradiation, and the temperature has no influence on the structure of the curcumin analogue under the condition of keeping out of the sun.
Further, the good solvent is preferably acetonitrile, methanol, ethanol, acetone, or tetrahydrofuran.
Furthermore, the isomer product with 30 percent of conversion rate can be obtained by UV irradiation for 2h, the preparation method is simple, and no by-product is generated.
Method for preparing cis-trans isomer Mixture M1(mix 1) of curcumin analogue
Preparing methanol solution containing 1mg/mL curcumin analogue, sealing with aluminum foil paper, and standing at 4 deg.C, 25 deg.C and 50 deg.C for 8 hr to obtain reaction product 1-3; and additionally preparing methanol solution of curcumin analogs with the same concentration, and respectively carrying out sunlight irradiation for 2h, sunlight irradiation for 24h, ultraviolet irradiation for 2h and radioactive iodine 131 irradiation for 2h at room temperature to obtain reaction products 4-7 so as to obtain cis-trans isomer mixtures of curcumin analogs in various proportions.
Results
(1) Molecular weight identification of the conversion products
The molecular weight of the product was determined by using a high performance liquid chromatography-time of flight mass spectrometer, as can be seen from fig. 20, the molecular weights of the curcumin analogues and the converted products thereof were 294.34, and both were confirmed to be isomers, and further, as can be seen from the structures, they were cis-trans isomers.
(2) Determination of the conversion of cis-trans isomers:
the cis-trans isomer conversion of curcumin analogs in sample solutions 1-7 was determined by High Performance Liquid Chromatography (HPLC) at a maximum absorption wavelength of 384 nm. The results are shown in fig. 21, and no new substance was produced in any of the reaction products 1 to 3, indicating that no isomer conversion of curcumin analogues occurred (fig. 21a to c). In the reaction product 4, 73.91% of the curcumin analogue content was converted into its isomer after being irradiated by sunlight for 2h (fig. 21 d); and after 24h of sunlight irradiation, the reaction product 5 has a plurality of complex products besides curcumin analogue isomers (figure 21 e). The conversion rate of the curcumin analogue isomer of the reaction product 6 after ultraviolet irradiation for 2h was 29.59% (fig. 21 f). The conversion rate of the curcumin analogue isomer of the reaction product 7 under the condition of radioactive iodine 131 radiation for 2h is 27.91% (fig. 21 g).
According to the test results of example 1, the product 6 (ultraviolet irradiation for 2h, isomer conversion rate of curcumin analogue is about 30%) which has simple and convenient preparation method and no byproduct generation is preferably selected as a representative mixed isomer M1 for subsequent biological activity study.
Example 2: preparation of curcumin analogue M1 nasal cavity nano preparation
The curcumin analog M1 used in this example was the product 6 prepared in example 1.
5mL of tetrahydrofuran solution containing 1mg/mL of M1 and 2mg/mL of carboxyl polyethylene glycol is prepared and mixed evenly, 200 mu L of M1 molecular solution is dropwise added into 5mL of deionized water, and nitrogen is blown while dropwise adding to remove the organic solvent. Stirring for 10min at 25 deg.C, standing to obtain M1 self-carried carrier-free nanoparticle suspension, and freeze drying to obtain lyophilized powder. Before use, the freeze-dried powder is re-dispersed in isotonic normal saline, the freeze-dried powder is re-dispersed in the isotonic normal saline, the freeze-dried powder is dropwise added into a chitosan oligosaccharide (0.1w/v) normal saline solution, and the mixture is stirred for 0.5 to 2 hours, so that the self-carrying type carrier-free M1 nasal nano preparation is obtained.
Results
(1) Determination of M1 nanoparticle morphology, particle size and potential distribution
The M1 nanoparticles prepared in example 1 were observed using a scanning electron microscope (FEI Quanta200, the netherlands) according to the method described in the specification thereof, and the scanning electron microscope image thereof is shown in fig. 1 a. The M1 nanoparticles prepared in example 1 were observed using a high-resolution transmission electron microscope (FEI Technai F30, the netherlands) according to the method described in the specification thereof, and the transmission electron microscope image thereof is shown in fig. 2 a. The M1 nanoparticles prepared in example 1 were subjected to dynamic light scattering measurements using a laser particle sizer (malvern, uk) according to the method described in the specification, and the M1 nanoparticles prepared in example 1 were found to have an average particle size of 62.73nm and a particle size distribution as shown in figure 3 a.
(2) Determination of M1 nanoparticle drug loading rate
M1 molecule prepared in example 1 was dissolved in acetonitrile, and a series of concentrations of M1 acetonitrile (6.25, 12.5, 25, 50 and 100ug/mL) was prepared in a gradient, and absorbance was measured at 428nm using Ultra Performance Liquid Chromatography (UPLC) to prepare a standard curve. And (3) taking three batches of 100ug/mL M1 nanoparticles, dissolving the nanoparticles in acetonitrile respectively, performing ultrasonic treatment for 5min, performing the same method for determination, and calculating the content of M1 in the nanoparticles by using a standard curve. As shown in fig. 4a, the drug loading rate of M1 nanoparticles was (31.49 ± 2.03)%.
(3) Surface potential measurement of M1 particles
Zeta-potential analysis of the M1 nanoparticles prepared in example 1 using a laser particle sizer according to the method described in the specification determined that the M1 nanoparticles prepared in example 1 had an average charge of-56.5 mV, with the distribution shown in FIG. 5.
(4) Optical characterization of M1 particles
When the M1 small molecule prepared in example 1 and the M1 nanoparticle prepared in example 1 were dissolved in water and organic solvent, respectively, as shown in fig. 6, it can be seen that the M1 small molecule is difficult to dissolve in water and soluble in tetrahydrofuran, while the M1 nanoparticle can be dispersed in water and has the tyndall effect under laser.
(5) M1 nanoparticle drug release profile determination
The M1 nanoparticles prepared in example 1 were equally divided into six parts, 3 parts were added to the artificial nasal solution, 3 parts were added to 5% plasma, which were separately filled into dialysis bags, dispersed and diluted, and separately added to dialysis bags (3500 molecular weight,
Figure GDA0003017689500000101
usa) followed by soaking in 200 ml of buffer of the same pH with constant stirring at 37 ℃ and collecting 2ml of solution from the solution at a certain time point. During dialysis, 2ml PBS was added after each sampling to keep the solution volume constant. And (4) measuring the absorbance by adopting a UV-VIS method, and calculating the drug release amount. Each sample was tested 3 times, averaged, and statistically analyzed, with the results shown in fig. 7. As can be seen, the M1 nanoparticles prepared in example 2The granules have slow release properties, do not show initial explosive drug release, but release slowly and stably, which is crucial for the application of M1 nanoparticles in vivo, and can reduce drug toxicity, drug leakage, etc.
(6) Cytotoxicity assays
The neuroma blast N2a cells were cultured according to the method described in the literature (cell culture, Sedrin Town, world book publishing Co., 1996), then the M1 nanoparticles prepared in example 1 were added to the cells and the cells were cultured, and after 24 hours of addition, the cell viability was measured according to the method described in the literature (MTT method), which is the M1 nanoparticle group. A group containing N2a cells treated with the same concentration of free M1 as the M1 nanoparticle group in the same manner was used as a positive control group; the negative control group was N2a cells cultured in a blank medium without the hydrophobic drug, wherein the survival rate of the cells in the negative control group was calculated as 100%. As shown in fig. 8, as the concentration increases, the free M1 can exhibit dose-dependent cytotoxicity, while the M1 nanoparticles in example 2 have no toxic effect on N2a cells at the same concentration, and may inhibit the cumulative toxicity of M1 small molecule drugs at higher concentrations due to the sustained release effect of M1 nanoparticles.
(7) Neuroprotective assay for M1 nanoparticles
The neural cell line PC12 cells were treated with MPP + neurotoxin to create a model of neurotoxic cells. Adding M1 nanoparticles prepared in example 1 for pretreatment 6h before molding to obtain a M1 nanoparticle group; model control group was obtained without drug treatment, and normal control group was obtained without MPP + neurotoxin. After modeling, the cells are continuously cultured for 48h, and the absorbance is measured according to a literature method, the result is shown in fig. 9, the cell survival rate of the M1 nanoparticle group is obviously higher than that of the MPP + model group, and the M1 nanoparticles prepared in example 2 can protect PC12 nerve cells in a dose-dependent manner and reduce cell damage induced by MPP + neurotoxin.
(8) Determination of binding Effect of M1 nanoparticles to target protein TFEB protein
The target protein of free M1 molecule for neuroprotection is TFEB protein in cytoplasm, and M1 can promote dephosphorylation of TFEB protein into nucleus and up-regulate expression of autophagy-related gene, thereby playing neuroprotection role. In this experiment, M1 nanoparticles prepared in example 2 were added to MF7 cells overexpressing a fluorescent-labeled TFEB protein, and after 24h of treatment, TFEB nuclear entry was observed, and as a result, as shown in fig. 10, M1 nanoparticles prepared in example 2 can promote TFEB nuclear entry in a dose-dependent manner, confirming that M1 nanoparticles retain the targeting property of the original molecule.
(9) Induction of autophagy flow by M1 nanoparticles
When autophagy is induced, the expression level of the marker protein LC3 is increased. The effect of drugs on autophagy flow can be examined under confocal fluorescence microscopy by constructing a lentivirus expressing GFP-RFP-LC3 and infecting N2a cells. Among them, the GFP-LC3 protein is a green acid-responsive protein and can be degraded in acid lysosomes, while the RFP-LC3 protein is a red acid-stable protein and cannot be degraded in lysosomes, so that when autophagy pathways are activated and autophagy flow is unobstructed, the number of red LC3 spots is increased, showing the induction of autophagy flow. In N2a cells infected with lentivirus, M1 nanoparticles prepared in example 1 were added for drug treatment to obtain M1 nanoparticle group, and after 24 hours, the number of red LC3 dots was measured under a confocal microscope, as shown in fig. 11, the number of red LC3 dots in the cells of the M1 nanoparticle group was significantly increased compared to the control group, confirming that M1 nanoparticles were able to induce autophagy flow. This is the linkage effect of the TFEB protein activated in the result of the test (8).
Example 3: m1 nasal cavity nano preparation brain targeting delivery system carrying fluorescent probe TPAAQ
TPAAQ is a hydrophobic micromolecule fluorescent probe excited by 473nm wavelength and emitted by 650nm wavelength, and can be used for monitoring in-vivo fluorescence distribution of nano materials. Because it is also a hydrophobic small molecule, similar to the preparation process of the M1 nanoparticles of example 1, M1 nasal cavity nano preparation carrying TPAAQ can be obtained by the same method.
5mL of tetrahydrofuran solution containing 1mg/mL of M1 and 2mg/mL of TPAAQ is prepared and mixed evenly, 200 mu L of the M1 molecular solution is added into 5mL of deionized water dropwise, and nitrogen is blown at the same time to remove the organic solvent. Magnetically stirring the mixture for 10 minutes at the temperature of 25 ℃, standing the mixture to obtain M1 self-carried carrier-free nanoparticle suspension emulsion carrying the fluorescent probe TPAAQ, and freeze-drying the suspension emulsion to form freeze-dried powder. Before use, the freeze-dried powder is re-dispersed in isotonic normal saline, added into the chitosan oligosaccharide (0.1w/v) normal saline solution drop by drop, stirred for 0.5-2h, reacted for 1 h by physical adsorption, centrifuged for 5-30min at the rotating speed of 10000-.
Results
(1) Determination of morphology and particle size distribution of M1 nanoparticles carrying fluorescent probe TPAAQ
The M1 nanoparticles prepared in example 1 were observed using a scanning electron microscope (FEI Quanta200, the netherlands) according to the method described in the specification, the scanning electron microscope image of which is shown in fig. 1 b. The M1 nanoparticles prepared in example 1 were observed using a high-resolution transmission electron microscope (FEI Technai F30, the netherlands) according to the method described in the specification thereof, and the transmission electron microscope image thereof is shown in fig. 2 b. The M1 nanoparticles prepared in example 1 were subjected to dynamic light scattering measurements using a laser particle sizer (malvern, uk) according to the method described in the specification, and the M1 nanoparticles prepared in example 1 were found to have an average particle size of 178.2nm, the particle size distribution chart being shown in figure 3 b.
(2) Determination of M1 nanoparticle drug loading rate of fluorescent probe TPAAQ
Using the standard curve of M1 acetonitrile solution prepared in the test (2) of example 1, three batches of 100ug/mL fluorescent probe TPAAQ-loaded M1 nanoparticles were dissolved in acetonitrile, and subjected to ultrasonic treatment for 5min, and the content of M1 in the nanoparticles was calculated using the standard curve. As shown in FIG. 4b, the drug loading rate of the M1 nanoparticles loaded with the fluorescent probe TPAAQ is (26.95. + -. 1.50)%.
(3) Cell uptake assay
Nerve cells are normally cultured, the M1 nasal nano preparation carrying the fluorescent probe TPAAQ prepared in example 3 is added, after 3 hours of culture, the cell uptake condition is observed under a laser confocal scanning microscope at a specific wavelength, as shown in figure 12, the M1 nasal nano preparation carrying the fluorescent probe TPAAQ prepared in example 3 can be greatly taken by the cells as seen from a fluorescent signal.
Example 4: application of M1 nanoparticles to nasal brain targeting delivery system
6 male C57BL/6J mice of strain 25g were selected and acclimatized for 3 days. The M1 nasal cavity nano preparation prepared in example 1 is dispersed in isotonic physiological saline with the concentration of 5mg/ml, 15ul of the nano preparation is given to a mouse nasal cavity, brain tissue, cerebrospinal fluid and plasma are dissected and taken out after 24h, the brain tissue is divided into an olfactory bulb part and the rest part of the brain, all samples are respectively added with methanol to remove protein, and the content of M1 drugs in the samples is analyzed by using triple quadrupole liquid chromatography-mass spectrometry. The results are shown in fig. 13, the M1 nasal nano-formulation brain-targeted delivery system delivers M1 drug into olfactory bulb with very high targeting, and has a distribution in cerebrospinal fluid that is more than three times the content in plasma, and a distribution in other parts of brain that is twice the amount of plasma. It was confirmed that the absorption pathway was through the olfactory bulb to the brain and was transmitted to other parts of the brain. Its delivery may be time-dependent and continues to pass back through the cerebrospinal fluid after 24 h.
Example 5: application of M1 nanoparticles loaded with TPAAQ fluorescent probe to nose-brain targeted delivery system
9 male C57BL/6J mice of strain 25g were selected and acclimatized for 3 days. The M1 nasal cavity nano preparation carrying the fluorescent probe TPAAQ prepared in the example 2 is dispersed in physiological saline with the concentration of 5mg/ml, 15ul of the nano preparation is given to a mouse nasal cavity, a small animal fluorescence imaging system is applied after 24h and 48h respectively, the in-vivo fluorescence of the brain of the mouse and fluorescence signals in organs and blood such as a brain, a heart, a liver, a spleen, a lung, a kidney and the like are detected, and the result is shown in fig. 14, the brain signals are remarkably stronger than other parts and tissues of a body, which indicates that the brain targeting delivery system in the example 3 can successfully deliver the M1 nasal cavity nano preparation into the brain with high targeting, and the distribution of the medicine in peripheral tissues is reduced.
Example 6: therapeutic application of self-carried carrier-free M1 nasal nano-preparation in Parkinson model mice
30 male C57BL/6J strain mice weighing 25g were divided into three groups, a first wild type group (WT group), a second model group (MPTP group), and a third model-administered group (M1 NPs), each of which was 10 mice. The second and third groups of mice were injected intraperitoneally with MPTP neurotoxin for five days at a dose of 20mg/kg following literature procedures to create a model of Parkinson's disease. The mice in WT group and MPTP group are administrated with normal saline in the nasal cavity, the mice in M1 NPs group are administrated with self-carried carrier-free M1 nasal cavity nano preparation in the nasal cavity, namely, the M1 nasal cavity nano preparation prepared in the example 1 is dispersed in isotonic normal saline and is prepared for clinical use, the concentration is 1mg/ml, and the mice are administrated with 15ul nasal cavity. The administration is carried out at intervals of one day, four times of administration are carried out, the behavioral observation is carried out after two weeks of molding, the mice are dissected afterwards, the brain tissue is separated, and various pharmacological tests are carried out.
Results
(1) Detection of behavior of Parkinson model mouse by open field test
The MPTP Parkinson mouse model has the symptoms of exploration dyskinesia, obvious anxiety and the like, and can be detected by an open field test. The behavioral performance of the parkinson model mice in example 5 was examined according to literature methods. The results are shown in fig. 15a, compared with the wild type mice in the control group, the movement track of the model mice is significantly changed, and the movement track of the model mice is approximately normal after the treatment of the M1 nasal cavity nano preparation. Statistical data show that compared with wild mice, the movement time (figure 15b), the average speed (figure 15c), the regional shuttling times (figure 15d) and the like of model mice in a mine are all remarkably reduced, and after the self-carried type carrier-free M1 nasal nano-preparation is used for treatment, the pathological changes are all remarkably improved, and the M1 nasal nano-preparation is proved to be capable of effectively relieving the behavioral symptoms of the Parkinson disease model.
(2) Gait test for detecting behavior expression of Parkinson model mouse
The clinical manifestations of Parkinson's disease mainly include resting tremor, bradykinesia, muscular rigidity, gait disorder of posture and the like. A DigiGait imaging system is adopted on an animal, the animal is imaged under a transparent running belt, and software quantifies characteristics such as gait mechanics, posture index and the like, so that the behavioral characteristics of the Parkinson model mouse can be detected. The results are shown in fig. 16, compared with the wild type mouse, the gait signal of the parkinson model mouse is disordered, the coordination is reduced, the sole contact area is obviously reduced, and after the treatment of the self-carrying type carrier-free M1 nasal cavity nano preparation, the pathological conditions are all obviously improved, and the M1 nasal cavity nano preparation is proved to be capable of effectively improving the behavioral symptoms of parkinson diseases.
(3) Tissue electron microscope detection of olfactory bulb, striatum and substantia nigra distribution of Parkinson model mice after nasal administration of M1 nanoparticles
Taking three mice in the treatment group, administering M1 nasal cavity nano preparation in the last nasal cavity for 24h, dissecting, taking out brain tissue, separating olfactory bulb, striatum and substantia nigra parts, fixing the section, and observing the distribution of each brain region of the nano particles under a transmission electron microscope. As shown in fig. 17, it is evident that M1 nanoparticles are distributed in the olfactory bulb, striatum and substantia nigra of brain, confirming that after M1 nasal nano-formulation is administered nasally, the nanoparticle can enter brain tissue as its prototype, while the olfactory bulb is distributed in the greatest amount, suggesting that its absorption pathway is mediated by olfactory nerves and spread backwards to other areas.
(4) Western Blot method for detecting toxic protein at brain olfactory bulb part and expression content of autophagy pathway related protein
Tyrosine Hydroxylase (TH) is a key enzyme in dopamine biosynthetic pathway, SNCA is an alpha-synuclein accumulated in brain of parkinson's disease, and TFEB is an autophagy pathway-related protein. After the administration of each group of mice was completed, the mice in the control group, the model group and the treatment group were sacrificed, brain tissues were taken, olfactory bulb sites were separated, the total amount of proteins was measured after tissue homogenization, and the expression amounts of the above proteins in the olfactory bulb homogenate were measured according to the conventional procedure of Western Blot using the above protein antibodies of Santa Cruz Co. As shown in fig. 18a, statistics indicate that TH protein is significantly decreased in the model group and significantly increased in the treatment group (fig. 18b), confirming that M1 nasal nano-formulation can alleviate dopamine synthesis disorder in brain caused by toxin. Compared with the control group and the model group, the amount of TFEB protein in the olfactory bulb of the brain of the treatment group is remarkably increased (fig. 18c), and it is confirmed that M1 nasal nano-preparation activates TFEB protein in the olfactory bulb, which is one of the possible mechanisms of the drug effect. Furthermore, SNCA toxic protein content increased in the model group, while the treatment group tended to decrease (fig. 18d), demonstrating that M1 nasal nano-formulation was able to eliminate toxic proteins in the olfactory bulb. In conclusion, after the M1 nano preparation is administrated through a nasal cavity, accumulation of neurotoxin in an olfactory bulb can be eliminated, dopamine synthesis disorder caused by toxin can be relieved, and the drug effect is probably related to M1 induction of an autophagy pathway mediated by TFEB protein.
(5) Western Blot method for detecting toxic protein at substantia nigra part of brain and expression content of autophagy pathway related protein
After the administration of each group of mice is finished, the mice of the control group, the model group and the treatment group are sacrificed, brain tissues are taken, olfactory bulb parts are separated, the total protein amount is measured after tissue homogenization, and the expression amount of the monomer alpha-synuclein and the aggregate alpha-synuclein in olfactory bulb homogenate is detected by applying an alpha-synuclein antibody of Santa Cruz according to the conventional steps of Western Blot. The result is shown in fig. 19a, statistics show that the monomer and aggregate alpha-synuclein are obviously increased in the model group, and the treatment group has a significantly reduced trend (fig. 19b and 19c), which proves that the M1 nasal nano-preparation can effectively eliminate toxic protein accumulation at the black lesion site, thereby having a therapeutic effect on diseases.
The applicant declares that the present invention is described by the above embodiments as the detailed features and the detailed methods of the present invention, but the present invention is not limited to the above detailed features and the detailed methods, that is, it is not meant that the present invention must be implemented by relying on the above detailed features and the detailed methods. It will be apparent to those skilled in the art that any modification of the present invention, equivalent substitutions of selected components of the present invention, additions of auxiliary components, selection of specific modes and the like, are within the scope and disclosure of the present invention.

Claims (10)

1. An autophagy inducer, which is characterized by comprising a cis isomer or a cis-trans isomer mixture of a hydrophobic small molecule curcumin analogue, wherein the cis isomer or the cis-trans isomer mixture is generated by irradiating the curcumin analogue with ultraviolet light for 1.5-2.5h, and the structural formula is as follows:
Figure 458170DEST_PATH_IMAGE001
2. an autophagy inducing agent according to claim 1, wherein the cis isomer or mixture of cis and trans isomers of curcumin analogs is obtained by subjecting a solution of curcumin analogs in methanol to ultraviolet irradiation for 1.5 to 2.5 hours.
3. A nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases is characterized in that: comprises hydrophobic small molecules with autophagy induction effect and amphiphilic surfactant; firstly, preparing 1-10mg/mL and 0.5-5mg/mL of a good solvent solution of hydrophobic small molecules with autophagy induction effects for an amphiphilic surfactant, and then dropwise adding the good solvent solution into deionized water, wherein the volume ratio of the good solvent solution to the deionized water is (0.5-5): 50, air blowing is assisted while dropwise adding, and good solvent volatilization is assisted; preparing self-carried carrier-free nano particle suspension emulsion with the particle size of 50-200nm by a reprecipitation method, and freeze-drying to prepare freeze-dried powder; before use, re-suspending the freeze-dried powder in isotonic physiological saline containing chitosan oligosaccharide to obtain the self-carried carrier-free nasal nano autophagy inducer; the concentration of the chitosan oligosaccharide in isotonic physiological saline solution is 0.01-0.2% (w/v);
wherein the hydrophobic small molecule with autophagy induction comprises cis-isomer or cis-trans-isomer mixture of curcumin analogues, wherein the cis-isomer or cis-trans-isomer mixture is generated by ultraviolet irradiation of the curcumin analogues with the following structural formula for 1.5-2.5 h:
Figure 109601DEST_PATH_IMAGE002
4. the nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases according to claim 3, wherein the early neurodegenerative diseases comprise Alzheimer's disease and Parkinson's disease.
5. The nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases according to claim 3, wherein the early neurodegenerative diseases are accompanied by symptoms of dysosmia, and the nasal cavity nano autophagy inducer is an autophagy inducer with high targeting enrichment at olfactory bulb parts.
6. The nasal nano autophagy inducer for preventing and treating early neurodegenerative diseases according to claim 3, characterized in that: the weight ratio of the cis-isomer in the mixture is 25-35% of the total mixture.
7. The nasal nano autophagy inducer for preventing and treating early neurodegenerative diseases according to claim 3, characterized in that: the mixture of cis-isomer accounting for 25-35% of the total mixture is prepared through ultraviolet irradiation of methanol solution of curcumin analog for 1.5-2.5 hr.
8. The nasal nano autophagy inducer for preventing and treating early neurodegenerative diseases according to claim 3, characterized in that: the amphiphilic surfactant is carboxyl polyethylene glycol or polymaleic anhydride 18 carbene-polyethylene glycol.
9. The nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases according to any one of claims 3 to 6, which is in the form of nasal spray or nasal drops.
10. A preparation method of a nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases comprises the following steps:
1) firstly, preparing a good solvent solution of carboxyl polyethylene glycol or polymaleic anhydride 18 carbene-polyethylene glycol 1-10mg/mL and 0.5-5mg/mL of hydrophobic micromolecule drug, and then dropwise adding the good solvent solution into deionized water: the volume ratio of the good solvent solution to the deionized water is (0.5-5): 50, blowing gas while dropwise adding to assist in volatilizing a good solvent; wherein the hydrophobic micromolecules comprise cis-isomer or cis-trans-isomer mixture of curcumin analogues, wherein the cis-isomer or cis-trans-isomer mixture is the curcumin analogues with the following structural formula, and is generated by ultraviolet irradiation for 1.5-2.5 h:
Figure 44059DEST_PATH_IMAGE002
2) preparing self-carried carrier-free nano particle suspension emulsion with the particle size of 50-200nm by a reprecipitation method, and freeze-drying to prepare freeze-dried powder;
3) before use, the freeze-dried powder is re-suspended in isotonic normal saline containing chitosan oligosaccharide to obtain the nasal cavity nano autophagy inducer, wherein the concentration of the chitosan oligosaccharide in the isotonic normal saline solution is 0.01-0.2% (w/v).
CN201910019951.6A 2019-01-09 2019-01-09 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof Active CN109731105B (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN201910019951.6A CN109731105B (en) 2019-01-09 2019-01-09 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof
CN202110665044.6A CN113384541B (en) 2019-01-09 2019-01-09 Nasal cavity nanometer autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof
PCT/CN2020/070987 WO2020143688A1 (en) 2019-01-09 2020-01-08 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method therefor
US17/372,403 US20220000804A1 (en) 2019-01-09 2021-07-09 Intranasal nano inducer for preventing and treating neurodegenerative diseases and method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910019951.6A CN109731105B (en) 2019-01-09 2019-01-09 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202110665044.6A Division CN113384541B (en) 2019-01-09 2019-01-09 Nasal cavity nanometer autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Publications (2)

Publication Number Publication Date
CN109731105A CN109731105A (en) 2019-05-10
CN109731105B true CN109731105B (en) 2021-06-04

Family

ID=66364126

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201910019951.6A Active CN109731105B (en) 2019-01-09 2019-01-09 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof
CN202110665044.6A Active CN113384541B (en) 2019-01-09 2019-01-09 Nasal cavity nanometer autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202110665044.6A Active CN113384541B (en) 2019-01-09 2019-01-09 Nasal cavity nanometer autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Country Status (3)

Country Link
US (1) US20220000804A1 (en)
CN (2) CN109731105B (en)
WO (1) WO2020143688A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113384541A (en) * 2019-01-09 2021-09-14 纳菲(深圳)制药科技有限公司 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112374976B (en) * 2020-11-24 2022-08-23 纳菲(深圳)制药科技有限公司 Novel method for synthesizing curcumin analogue

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH10503596A (en) * 1995-05-23 1998-03-31 フィリップス エレクトロニクス ネムローゼ フェンノートシャップ Radiation dose indicator, lamp and tanning device with such radiation dose indicator
US8841326B2 (en) * 2004-02-12 2014-09-23 Stc.Unm Therapeutic curcumin derivatives
CN102512404B (en) * 2011-11-18 2014-05-07 上海交通大学 Lung targeting preparation of curcumin class compound as well as preparation method and application thereof
US9351946B2 (en) * 2014-03-06 2016-05-31 Hong Kong Baptist University MTOR-independent activator of TFEB for autophagy enhancement and uses thereof
CN104288151B (en) * 2014-10-16 2017-06-23 河南工业大学 Applications of the curcumin derivate DIMMO in novel autophagy derivant is prepared
CN109010846B (en) * 2017-12-27 2021-11-02 湖南师范大学 Polyethylene glycol-chitosan-curcumin polymer, drug-loaded nanoparticles thereof and preparation method
CN109731105B (en) * 2019-01-09 2021-06-04 纳菲(深圳)制药科技有限公司 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113384541A (en) * 2019-01-09 2021-09-14 纳菲(深圳)制药科技有限公司 Nasal cavity nano autophagy inducer for preventing and treating early neurodegenerative diseases and preparation method thereof

Also Published As

Publication number Publication date
CN113384541B (en) 2023-05-23
CN113384541A (en) 2021-09-14
CN109731105A (en) 2019-05-10
US20220000804A1 (en) 2022-01-06
WO2020143688A1 (en) 2020-07-16

Similar Documents

Publication Publication Date Title
Formica et al. On a highway to the brain: A review on nose-to-brain drug delivery using nanoparticles
Haque et al. Development and evaluation of brain targeted intranasal alginate nanoparticles for treatment of depression
Ray et al. Polysorbate 80 coated crosslinked chitosan nanoparticles of ropinirole hydrochloride for brain targeting
Musumeci et al. Oxcarbazepine free or loaded PLGA nanoparticles as effective intranasal approach to control epileptic seizures in rodents
Correia et al. Lipid nanoparticles strategies to modify pharmacokinetics of central nervous system targeting drugs: Crossing or circumventing the blood–brain barrier (BBB) to manage neurological disorders
Zhang et al. Preparation and characterization of insulin-loaded bioadhesive PLGA nanoparticles for oral administration
US20190117673A1 (en) Ganaxolone formulation and use thereof
Gao et al. Brain delivery of vasoactive intestinal peptide enhanced with the nanoparticles conjugated with wheat germ agglutinin following intranasal administration
Alam et al. Development and evaluation of thymoquinone-encapsulated chitosan nanoparticles for nose-to-brain targeting: a pharmacoscintigraphic study
Wilson et al. Chitosan nanoparticles as a new delivery system for the anti-Alzheimer drug tacrine
Wu et al. A novel small Odorranalectin-bearing cubosomes: preparation, brain delivery and pharmacodynamic study on amyloid-β25–35-treated rats following intranasal administration
US20050175668A1 (en) Lapachone delivery systems, compositions and uses related thereto
WO2020143662A1 (en) Brain-targeted delivery system for carrier-free nasal nano preparation modified by chitosan oligosaccharide and preparation method therefor
US20220000804A1 (en) Intranasal nano inducer for preventing and treating neurodegenerative diseases and method thereof
Ourani-Pourdashti et al. Preparation and evaluation of niosomal chitosan-based in situ gel formulation for direct nose-to-brain methotrexate delivery
Vallorz et al. Formoterol PLGA-PEG nanoparticles induce mitochondrial biogenesis in renal proximal tubules
Wei et al. A novel delivery method of cyclovirobuxine D for brain-targeting: chitosan coated nanoparticles loading cyclovirobuxine D by intranasal administration
Khemariya et al. New-fangled approach in the management of Alzheimer by formulation of polysorbate 80 coated chitosan nanoparticles of rivastigmine for brain delivery and their in vivo evaluation
CN115667214A (en) Nanoparticles comprising drug dimers and uses thereof
US11534410B2 (en) Amphotericin loaded PEGylated lipid nanoparticles and methods of use
Zhang et al. Non-invasive drug delivery systems mediated by nanocarriers and molecular dynamics simulation for posterior eye disease therapeutics: Virtual screening, construction and comparison
Pavlov et al. Study of adhesive properties of new dosage forms for Nano-L-DOPA nasal delivery system based on PLGA nanoparticles
Pavlov et al. The Research of Physicochemical Properties and Determination of Nano-L-DOPA Quality Attributes Based on PLGA Nanoparticles for the Treatment of Parkinson's Disease
Joseph et al. Development, characterization and evaluation of antiretroviral drug–Didanosine loaded serum albumin nanocarriers for an antiretroviral therapy
Bhatt et al. Formulation and Evaluation of Brain-Targeted Nanocarrier of Sertraline Hydrochloride via Intranasal Administration

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20191129

Address after: 518118 workshop b4401, Life Science Park, Shencheng investment creative factory, JuLongshan a road, xiuxin community, Kengzi street, Pingshan District, Shenzhen City, Guangdong Province

Applicant after: Nafi (Shenzhen) Pharmaceutical Technology Co., Ltd

Address before: Xiamen City, Fujian Province, 361000 South Siming Road No. 422

Applicant before: Xiamen University

TA01 Transfer of patent application right
GR01 Patent grant
GR01 Patent grant